Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 285(31): 23818-28, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20504765

RESUMO

Nuclear translocation of chloride intracellular channel protein CLIC4 is essential for its role in Ca(2+)-induced differentiation, stress-induced apoptosis, and modulating TGF-beta signaling in mouse epidermal keratinocytes. However, post-translational modifications on CLIC4 that govern nuclear translocation and thus these activities remain to be elucidated. The structure of CLIC4 is dependent on the redox environment, in vitro, and translocation may depend on reactive oxygen and nitrogen species in the cell. Here we show that NO directly induces nuclear translocation of CLIC4 that is independent of the NO-cGMP pathway. Indeed, CLIC4 is directly modified by NO through S-nitrosylation of a cysteine residue, as measured by the biotin switch assay. NO enhances association of CLIC4 with the nuclear import proteins importin alpha and Ran. This is likely a result of the conformational change induced by S-nitrosylated CLIC4 that leads to unfolding of the protein, as exhibited by CD spectra analysis and trypsinolysis of the modified protein. Cysteine mutants of CLIC4 exhibit altered nitrosylation, nuclear residence, and stability, compared with the wild type protein likely as a consequence of altered tertiary structure. Moreover, tumor necrosis factor alpha-induced nuclear translocation of CLIC4 is dependent on nitric-oxide synthase activity. Inhibition of nitric-oxide synthase activity inhibits tumor necrosis factor alpha-induced nitrosylation and association with importin alpha and Ran and ablates CLIC4 nuclear translocation. These results suggest that S-nitrosylation governs CLIC4 structure, its association with protein partners, and thus its intracellular distribution.


Assuntos
Transporte Ativo do Núcleo Celular , Canais de Cloreto/química , Proteínas Mitocondriais/química , Nitrogênio/química , Animais , Diferenciação Celular , Canais de Cloreto/metabolismo , Queratinócitos/citologia , Camundongos , Proteínas Mitocondriais/metabolismo , Mutação , Células NIH 3T3 , Óxido Nítrico Sintase/metabolismo , Oxirredução , Fator de Necrose Tumoral alfa/metabolismo , alfa Carioferinas/metabolismo , Proteína ran de Ligação ao GTP/metabolismo
2.
J Cell Sci ; 120(Pt 15): 2631-40, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17636002

RESUMO

Keratinocyte differentiation requires integrating signaling among intracellular ionic changes, kinase cascades, sequential gene expression, cell cycle arrest, and programmed cell death. We now show that Cl(-) intracellular channel 4 (CLIC4) expression is increased in both mouse and human keratinocytes undergoing differentiation induced by Ca(2+), serum and the protein kinase C (PKC)-activator, 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Elevation of CLIC4 is associated with signaling by PKCdelta, and knockdown of CLIC4 protein by antisense or shRNA prevents Ca(2+)-induced keratin 1, keratin 10 and filaggrin expression and cell cycle arrest in differentiating keratinocytes. CLIC4 is cytoplasmic in actively proliferating keratinocytes in vitro, but the cytoplasmic CLIC4 translocates to the nucleus in keratinocytes undergoing growth arrest by differentiation, senescence or transforming growth factor beta (TGFbeta) treatment. Targeting CLIC4 to the nucleus of keratinocytes via adenoviral transduction increases nuclear Cl(-) content and enhances expression of differentiation markers in the absence of elevated Ca(2+). In vivo, CLIC4 is localized to the epidermis in mouse and human skin, where it is predominantly nuclear in quiescent cells. These results suggest that CLIC4 participates in epidermal homeostasis through both alterations in the level of expression and subcellular localization. Nuclear CLIC4, possibly by altering the Cl(-) and pH of the nucleus, contributes to cell cycle arrest and the specific gene expression program associated with keratinocyte terminal differentiation.


Assuntos
Cálcio/metabolismo , Diferenciação Celular , Canais de Cloreto/metabolismo , Queratinócitos/citologia , Proteína Quinase C/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Canais de Cloreto/isolamento & purificação , Proteínas Filagrinas , Expressão Gênica , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Queratinócitos/metabolismo , Queratinas/metabolismo , Camundongos , Isoformas de Proteínas/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição AP-1/metabolismo
3.
J Biol Chem ; 282(41): 29987-97, 2007 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-17656367

RESUMO

The divergent response and the molecular mechanisms underlying the anti-cancer effects of retinoid X receptor (RXR) ligand (rexinoid) therapy are poorly understood. This study demonstrates that ligand-activated RXR homodimer facilitated G(1) arrest by up-regulation of p21 in vitro and in vivo but failed to induce G(1) arrest when p21 expression was blocked by p21 small interfering RNA. RXR ligand-dependent p21 up-regulation was transcriptionally controlled through the direct binding of RXR homodimers to two consecutive retinoid X response elements in the p21 promoter. Structural overlap of a retinoic acid response element with these retinoid X response elements led to a high affinity binding of retinoic acid receptor/RXR heterodimer to the retinoic acid response element, resulting in the prevention of RXR ligand-mediated p21 transactivation. These data show that p21 is a potential and novel molecular target for RXR ligand-mediated anti-cancer therapy and that the expression level of retinoic acid receptor and RXR in tumors may be crucial to induce p21-mediated cell growth arrest in RXR ligand therapy.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Receptores do Ácido Retinoico/metabolismo , Transcrição Gênica , Animais , Ciclo Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Dimerização , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Ligação Proteica , Receptores X de Retinoides/metabolismo , Ativação Transcricional
4.
Mol Carcinog ; 46(8): 599-604, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17443730

RESUMO

Chloride intracellular channel 4 (CLIC4) is a putative chloride channel for intracellular organelles. CLIC4 has biological activities in addition to or because of its channel activity. In keratinocytes, CLIC4 resides in the mitochondria and cytoplasm, and CLIC4 gene expression is regulated by p53, TNF-alpha, and c-Myc. Cytoplasmic CLIC4 translocates to the nucleus in response to cellular stress conditions including DNA damage, metabolic inhibition, senescence, and exposure to certain trophic factors such as TNF-alpha and LPS. Nuclear translocation is associated with growth arrest or apoptosis, depending on the level of expression. In the nucleus CLIC4 interacts with several nuclear proteins as demonstrated by yeast two-hybrid screening and co-immunoprecipitation. Nuclear CLIC4 appears to act on the TGF-beta pathway, and TGF-beta also causes CLIC4 nuclear translocation. In human and mouse cancer cell lines, CLIC4 levels are reduced, and CLIC4 is excluded from the nucleus. CLIC4 soluble or membrane-inserted status is dependent on redox state, and redox alterations in cancer cells could underly the defect in nuclear translocation. CLIC4 is reduced and excluded from the nucleus of many human epithelial neoplasms. Paradoxically, CLIC4 is reciprocally upregulated in tumor stroma in conjunction with the expression of alpha-smooth muscle actin in the fibroblast to myofibroblast transition. Overexpression of CLIC4 in cancer cells inhibits tumor growth in vivo. Conversely, overexpression of CLIC4 in tumor stromal cells stimulates tumor growth in vivo. Thus, CLIC4 participates in normal and pathological processes and may serve as a useful target for therapies in disturbances of homeostasis and neoplastic transformation.


Assuntos
Canais de Cloreto/metabolismo , Regulação Neoplásica da Expressão Gênica , Homeostase , Queratinócitos/metabolismo , Neoplasias Cutâneas/metabolismo , Animais , Diferenciação Celular , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Humanos , Oligonucleotídeos Antissenso/farmacologia , Neoplasias Cutâneas/patologia
5.
Clin Cancer Res ; 13(1): 121-31, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17200346

RESUMO

PURPOSE: CLIC4, a member of a family of intracellular chloride channels, is regulated by p53, c-Myc, and tumor necrosis factor-alpha. Regulation by factors involved in cancer pathogenesis, together with the previously shown proapoptotic activity of CLIC4, suggests that the protein may have a tumor suppressor function. To address this possibility, we characterized the expression profile, subcellular localization, and gene integrity of CLIC4 in human cancers and determined the functional consequences of CLIC4 expression in tumor epithelium and stromal cells. EXPERIMENTAL DESIGN: CLIC4 expression profiles were analyzed by genomics, proteomics, bioinformatics, and tissue microarrays. CLIC4 expression, as a consequence of crosstalk between stroma and epithelium, was tested in vitro by coculture of breast epithelial tumor cells and normal fibroblasts, and the functional consequences of CLIC4 expression was tested in vivo in xenografts of human breast tumor cell lines reconstituted with CLIC4 or mixed with fibroblasts that overexpress CLIC4 transgenically. RESULTS: In cDNA arrays of matched human normal and tumor tissues, CLIC4 expression was reduced in renal, ovarian, and breast cancers. However, CLIC4 protein levels were variable in tumor lysate arrays. Transcript sequences of CLIC4 from the human expressed sequence tag database and manual sequencing of cDNA from 60 human cancer cell lines (NCI60) failed to reveal deletion or mutations in the CLIC4 gene. On matched tissue arrays, CLIC4 was predominantly nuclear in normal human epithelial tissues but not cancers. With advancing malignant progression, CLIC4 staining became undetectable in tumor cells, but expression increased in stromal cells coincident with up-regulation of alpha-smooth muscle actin, suggesting that CLIC4 is up-regulated in myofibroblasts. Coculture of cancer cells and fibroblasts induced the expression of both CLIC4 and alpha-smooth muscle actin in fibroblasts adjacent to tumor nests. Introduction of CLIC4 or nuclear targeted CLIC4 via adenovirus into human breast cancer xenografts inhibited tumor growth, whereas overexpression of CLIC4 in stromal cells of xenografts enhanced tumor growth. CONCLUSION: Loss of CLIC4 in tumor cells and gain in tumor stroma is common to many human cancers and marks malignant progression. Up-regulation of CLIC4 in tumor stroma is coincident with myofibroblast conversion, generally a poor prognostic indicator. Reactivation and restoration of CLIC4 in tumor cells or the converse in tumor stromal cells could provide a novel approach to inhibit tumor growth.


Assuntos
Canais de Cloreto/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias/patologia , Regulação para Cima , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Canais de Cloreto/genética , Análise Mutacional de DNA , Progressão da Doença , Epitélio/metabolismo , Fibroblastos/metabolismo , Genes Supressores de Tumor , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo
6.
J Biol Chem ; 281(5): 2750-6, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16316993

RESUMO

Myc is a key regulatory protein in higher eukaryotes controlling important cellular functions such as proliferation, differentiation, and apoptosis. Myc is profoundly involved in the genesis of many human and animal cancers, and the abrogation of Myc-induced apoptosis is a critical event in cancer progression. Because the mechanisms that mediate Myc-induced apoptosis are largely unknown, we analyzed protein expression during Myc-induced apoptosis using an isotope-coded affinity tag quantitative proteomics approach and identified that a proapoptotic mitochondrial chloride ion channel, mtCLIC/CLIC4, is induced by Myc. Myc binds to the mtCLIC gene promoter and activates its transcription. Suppression of mtCLIC expression by RNA interference inhibited Myc-induced apoptosis in response to different stress conditions and abolished the cooperative induction of apoptosis by Myc and Bax. We also found that Myc reduces the expression of Bcl-2 and Bcl-xL and that the apoptosis-inducing stimuli up-regulate Bax expression. These results suggest that up-regulation of mtCLIC, together with a reduction in Bcl-2 and Bcl-xL, sensitizes Myc-expressing cells to the proapoptotic action of Bax.


Assuntos
Apoptose/genética , Canais de Cloreto/genética , Proteômica , Proteínas Proto-Oncogênicas c-myc/fisiologia , Proteínas Reguladoras de Apoptose/genética , Regulação da Expressão Gênica , Humanos , Proteínas Mitocondriais/genética , RNA Interferente Pequeno/farmacologia , Estresse Fisiológico/genética , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/fisiologia
7.
J Investig Dermatol Symp Proc ; 10(2): 105-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16358817

RESUMO

Chloride intracellular channel (CLIC)4 is a p53- and tumor necrosis factor alpha (TNFalpha)-regulated chloride channel protein that is localized to the mitochondria and cytoplasm of mouse and human keratinocytes. CLIC4 protein increases in differentiating keratinocytes and in keratinocytes exposed to DNA-damaging agents and metabolic inhibitors. Increasing CLIC4 levels by transduction of recombinant CLIC4 causes apoptosis. CLIC4 translocates to the nucleus under a variety of conditions of cell stress, and nuclear CLIC4 is associated with cell cycle arrest and accelerated apoptosis. Reduction of CLIC4 and several other CLIC family members by expressing a doxycycline-regulated CLIC4 antisense also causes apoptosis in squamous cancer cell lines. Expressing antisense CLIC4 in tumors derived from transplanting these cells into nude mice inhibits tumor growth, increases tumor apoptosis, and reduces tumor cell proliferation. Co-administration of TNFalpha intraperitoneally enhances the tumor-inhibitory influence of CLIC4 antisense expression. Together, these results suggest that CLIC4 is important for keratinocyte viability and may be a novel target for anti-cancer therapy.


Assuntos
Canais de Cloreto/fisiologia , Neoplasias/terapia , Animais , Diferenciação Celular , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Humanos , Queratinócitos/citologia , Camundongos , Oligonucleotídeos Antissenso/farmacologia
8.
Curr Pharm Des ; 11(21): 2753-64, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16101453

RESUMO

The passage of ions to form and maintain electrochemical gradients is a key element for regulating cellular activities and is dependent on specific channel proteins or complexes. Certain ion channels have been the targets of pharmaceuticals that have had impact on a variety of cardiovascular and neurological diseases. Chloride channels regulate the movement of a major cellular anion, and in so doing they in part determine cell membrane potential, modify transepithelial transport, and maintain intracellular pH and cell volume. There are multiple families of chloride channel proteins, and respiratory, neuromuscular, and renal dysfunction may result from mutations in specific family members. Interest in chloride channels related to cancer first arose when the multidrug resistance protein (MDR/P-glycoprotein) was linked to volume-activated chloride channel activity in cancer cells from patients undergoing chemotherapy. More recently, CLC, CLIC, and CLCA intracellular chloride channels have been recognized for their contributions in modifying cell cycle, apoptosis, cell adhesion, and cell motility. Moreover, advances in structural biology and high-throughput screening provide a platform to identify chemical compounds that modulate the activities of intracellular chloride channels thereby influencing chloride ion transport and altering cell behavior. This review will focus on several chloride channel families that may contribute to the cancer phenotype and suggest how they may serve as novel targets for primary cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Canais de Cloreto/efeitos dos fármacos , Canais de Cloreto/fisiologia , Sequência de Aminoácidos , Animais , Cálcio/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Canais de Cloreto/classificação , Canais de Cloreto/genética , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Humanos , Dados de Sequência Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/fisiopatologia
9.
Cancer Res ; 65(2): 562-71, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15695400

RESUMO

mtCLIC/CLIC4 is a p53 and tumor necrosis factor alpha (TNFalpha) regulated intracellular chloride channel protein that localizes to cytoplasm and organelles and induces apoptosis when overexpressed in several cell types of mouse and human origin. CLIC4 is elevated during TNFalpha-induced apoptosis in human osteosarcoma cell lines. In contrast, inhibition of NFkappaB results in an increase in TNFalpha-mediated apoptosis with a decrease in CLIC4 protein levels. Cell lines expressing an inducible CLIC4-antisense construct that also reduces the expression of several other chloride intracellular channel (CLIC) family proteins were established in the human osteosarcoma lines SaOS and U2OS cells and a malignant derivative of the mouse squamous papilloma line SP1. Reduction of CLIC family proteins by antisense expression caused apoptosis in these cells. Moreover, CLIC4-antisense induction increased TNFalpha-mediated apoptosis in both the SaOS and U2OS derivative cell lines without altering TNFalpha-induced NFkappaB activity. Reducing CLIC proteins in tumor grafts of SP1 cells expressing a tetracycline-regulated CLIC4-antisense substantially inhibited tumor growth and induced tumor apoptosis. Administration of TNFalpha i.p. modestly enhanced the antitumor effect of CLIC reduction in vivo. These results suggest that CLIC proteins could serve as drug targets for cancer therapy, and reduction of CLIC proteins could enhance the activity of other anticancer drugs.


Assuntos
Apoptose/fisiologia , Neoplasias Ósseas/patologia , Canais de Cloreto/antagonistas & inibidores , DNA Antissenso/genética , NF-kappa B/antagonistas & inibidores , Osteossarcoma/patologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/terapia , Bovinos , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Canais de Cloreto/genética , Humanos , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , Osteossarcoma/genética , Osteossarcoma/terapia , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mod Pathol ; 18(4): 528-34, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15502807

RESUMO

The role of human papilloma virus (HPV) infection in the development of cervical carcinoma is well established, however, the prevalence of HPV DNA in cervical adenocarcinoma varies from study to study. It appears to be caused by a number of factors, one of which is that cervical adenocarcinomas comprise a heterogeneous group of multiple subtypes. To clarify the impact of HPV infection on the development of cervical adenocarcinoma with diverse histological subtypes, we performed a population-based study in Korean women from 15 different institutes for the status of HPV infection in adenocarcinoma of uterine cervix. A total of 432 cervical adenocarcinomas from 1997 to 2001 were reviewed and classified according to the modified WHO classification. For 135 cases, HPV typing was performed with HPV DNA chip (82 cases) and PCR HPV typing (53 cases), using formalin-fixed, paraffin-embedded archival tissue. The overall prevalence of HPV infection in cervical adenocarcinoma was 90%. The infection of HPV 16 and/or HPV 18 accounted for 78% of HPV-positive adenocarcinomas. Multiple HPV types were found in 13% of the cases. The HPV DNA was rarely detected in minimal deviation adenocarcinoma. Interestingly, HPV 16 was a predominant type in endometrioid and villoglandular types, whereas HPV 16 and HPV 18 were detected with equal prevalence in other subtypes. In conclusion, HPV infection, mostly HPV 16 and HPV 18, is highly associated with most of the cervical adenocarcinomas, whereas endometrioid and villoglandular type have a different pattern of HPV infection status. Minimal deviation adenocarcinoma does not seem to be related with HPV infection.


Assuntos
Adenocarcinoma/patologia , DNA Viral/genética , Papillomaviridae/genética , Infecções por Papillomavirus/patologia , Neoplasias do Colo do Útero/patologia , Adenocarcinoma/virologia , Adulto , DNA Viral/análise , Feminino , Genótipo , Humanos , Coreia (Geográfico)/epidemiologia , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Papillomaviridae/crescimento & desenvolvimento , Infecções por Papillomavirus/epidemiologia , Infecções por Papillomavirus/virologia , Prevalência , Neoplasias do Colo do Útero/virologia
11.
J Biol Chem ; 279(6): 4632-41, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14610078

RESUMO

CLIC4/mtCLIC, a chloride intracellular channel protein, localizes to the mitochondria and cytoplasm of keratinocytes and participates in the apoptotic response to stress. We now show that multiple stress inducers cause the translocation of cytoplasmic CLIC4 to the nucleus. Immunogold electron microscopy and confocal analyses indicate that nuclear CLIC4 is detected prior to the apoptotic phenotype. CLIC4 associates with the Ran, NTF2, and Importin-alpha nuclear import complexes in immunoprecipitates of lysates from cells treated with apoptotic/stress-inducing agents. Deletion or mutation of the nuclear localization signal in the C terminus of CLIC4 eliminates nuclear translocation, whereas N terminus deletion enhances nuclear localization. Targeting CLIC4 to the nucleus via adenoviral transduction accelerates apoptosis when compared with cytoplasmic CLIC4, and only nuclear-targeted CLIC4 causes apoptosis in Apaf null mouse fibroblasts or in Bcl-2-overexpressing keratinocytes. These results indicate that CLIC4 nuclear translocation is an integral part of the cellular response to stress and may contribute to the initiation of nuclear alterations that are associated with apoptosis.


Assuntos
Apoptose/fisiologia , Canais de Cloreto/metabolismo , Proteínas Mitocondriais/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Sequência de Bases , Células Cultivadas , Canais de Cloreto/genética , DNA/genética , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Proteínas Mitocondriais/genética , Mutagênese Sítio-Dirigida , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Organelas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência
12.
Genomics ; 82(1): 57-67, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12809676

RESUMO

Protein kinase C-delta (PKC-delta) is a ubiquitously expressed kinase involved in a variety of cellular signaling pathways including cell growth, differentiation, apoptosis, tumor promotion, and carcinogenesis. While signaling pathways downstream of PKC-delta are well studied, the regulation of the gene has not been extensively analyzed. A mouse genomic DNA fragment containing the PKC-delta gene was sequenced by the primer-walking method, and the subsequent DNA sequence data were used as a query to clone Caenorhabditis elegans and human genomic homologs from the publicly available genomic databases. The genomic structures of C. elegans, mouse, rat, and human PKC-delta were analyzed, and the result revealed that PKC-delta genes comprise 12, 18, 19, and 18 exons for C. elegans, mouse, rat, and human, respectively. The translation start methionine resides in the second exon in mouse and human and in the third exon in rat. The first intron between the first exon and the exon with the translation start methionine in mammalian genes represents a very large gap, as long as 17 kb in human, indicating a complexity involved in gene splicing. Overall exon-intron genomic structure is highly conserved among mammals, while significantly diverged in C. elegans. Putative transcription factor binding sites on the 1.7-kb promoter region of the mouse gene suggest that PKC-delta might be involved in spermatogenesis, embryogenesis, development, brain generation, immune response, oxidative environment, and oncogenesis. Studies on the promoter and subsequent biological testing on mouse keratinocytes indicate that tumor necrosis factor (TNF)-alpha increases the expression of PKC-delta, and this correlates with the time of NFkappaB nuclear translocation and activation. This TNF-alpha-mediated upregulation of PKC-delta is repressed in keratinocytes that are preinfected with IkappaB superrepressor adenovirus, suggesting that NFkappaB is involved directly in PKC-delta expression.


Assuntos
Genes/genética , Isoenzimas/genética , Regiões Promotoras Genéticas , Proteína Quinase C/genética , Animais , Animais Recém-Nascidos , Sequência de Bases , Caenorhabditis elegans/genética , Células Cultivadas , Evolução Molecular , Éxons , Genoma , Genoma Humano , Humanos , Íntrons , Isoenzimas/metabolismo , Queratinócitos/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase C/metabolismo , Ratos , Sequências Reguladoras de Ácido Nucleico , Análise de Sequência de DNA , Especificidade da Espécie , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/fisiologia
13.
Mol Cell Biol ; 22(11): 3610-20, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11997498

RESUMO

mtCLIC/CLIC4 (referred to here as mtCLIC) is a p53- and tumor necrosis factor alpha-regulated cytoplasmic and mitochondrial protein that belongs to the CLIC family of intracellular chloride channels. mtCLIC associates with the inner mitochondrial membrane. Dual regulation of mtCLIC by two stress response pathways suggested that this chloride channel protein might contribute to the cellular response to cytotoxic stimuli. DNA damage or overexpression of p53 upregulates mtCLIC and induces apoptosis. Overexpression of mtCLIC by transient transfection reduces mitochondrial membrane potential, releases cytochrome c into the cytoplasm, activates caspases, and induces apoptosis. mtCLIC is additive with Bax in inducing apoptosis without a physical association of the two proteins. Antisense mtCLIC prevents the increase in mtCLIC levels and reduces apoptosis induced by p53 but not apoptosis induced by Bax, suggesting that the two proapoptotic proteins function through independent pathways. Our studies indicate that mtCLIC, like Bax, Noxa, p53AIP1, and PUMA, participates in a stress-induced death pathway converging on mitochondria and should be considered a target for cancer therapy through genetic or pharmacologic approaches.


Assuntos
Apoptose/fisiologia , Canais de Cloreto/metabolismo , Dano ao DNA , Proteínas Proto-Oncogênicas c-bcl-2 , Proteína Supressora de Tumor p53/metabolismo , Animais , Sítios de Ligação/genética , Células Cultivadas , Canais de Cloreto/genética , Expressão Gênica , Genes p53 , Queratinócitos/citologia , Queratinócitos/metabolismo , Potenciais da Membrana , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transfecção , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA